1
|
Hu Y, Tao W. Current perspectives on microglia-neuron communication in the central nervous system: Direct and indirect modes of interaction. J Adv Res 2024; 66:251-265. [PMID: 38195039 PMCID: PMC11674795 DOI: 10.1016/j.jare.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 10/05/2023] [Accepted: 01/06/2024] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND The incessant communication that takes place between microglia and neurons is essential the development, maintenance, and pathogenesis of the central nervous system (CNS). As mobile phagocytic cells, microglia serve a critical role in surveilling and scavenging the neuronal milieu to uphold homeostasis. AIM OF REVIEW This review aims to discuss the various mechanisms that govern the interaction between microglia and neurons, from the molecular to the organ system level, and to highlight the importance of these interactions in the development, maintenance, and pathogenesis of the CNS. KEY SCIENTIFIC CONCEPTS OF REVIEW Recent research has revealed that microglia-neuron interaction is vital for regulating fundamental neuronal functions, such as synaptic pruning, axonal remodeling, and neurogenesis. The review will elucidate the intricate signaling pathways involved in these interactions, both direct and indirect, to provide a better understanding of the fundamental mechanisms of brain function. Furthermore, gaining insights into these signals could lead to the development of innovative therapies for neural disorders.
Collapse
Affiliation(s)
- Yue Hu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 220023, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weiwei Tao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 220023, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
2
|
Harackiewicz O, Grembecka B. The Role of Microglia and Astrocytes in the Pathomechanism of Neuroinflammation in Parkinson's Disease-Focus on Alpha-Synuclein. J Integr Neurosci 2024; 23:203. [PMID: 39613467 DOI: 10.31083/j.jin2311203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/04/2024] [Accepted: 07/31/2024] [Indexed: 12/01/2024] Open
Abstract
Glial cells, including astrocytes and microglia, are pivotal in maintaining central nervous system (CNS) homeostasis and responding to pathological insults. This review elucidates the complex immunomodulatory functions of glial cells, with a particular focus on their involvement in inflammation cascades initiated by the accumulation of alpha-synuclein (α-syn), a hallmark of Parkinson's disease (PD). Deriving insights from studies on both sporadic and familial forms of PD, as well as animal models of PD, we explore how glial cells contribute to the progression of inflammation triggered by α-syn aggregation. Additionally, we analyze the interplay between glial cells and the blood-brain barrier (BBB), highlighting the role of these cells in maintaining BBB integrity and permeability in the context of PD pathology. Furthermore, we delve into the potential activation of repair and neuroprotective mechanisms mediated by glial cells amidst α-syn-induced neuroinflammation. By integrating information on sporadic and familial PD, as well as BBB dynamics, this review aims to deepen our understanding of the multifaceted interactions between glial cells, α-syn pathology, and CNS inflammation, thereby offering valuable insights into therapeutic strategies for PD and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Oliwia Harackiewicz
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdańsk, 80-308 Gdańsk, Poland
| | - Beata Grembecka
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdańsk, 80-308 Gdańsk, Poland
| |
Collapse
|
3
|
Roodveldt C, Bernardino L, Oztop-Cakmak O, Dragic M, Fladmark KE, Ertan S, Aktas B, Pita C, Ciglar L, Garraux G, Williams-Gray C, Pacheco R, Romero-Ramos M. The immune system in Parkinson's disease: what we know so far. Brain 2024; 147:3306-3324. [PMID: 38833182 PMCID: PMC11449148 DOI: 10.1093/brain/awae177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/02/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024] Open
Abstract
Parkinson's disease is characterized neuropathologically by the degeneration of dopaminergic neurons in the ventral midbrain, the accumulation of α-synuclein (α-syn) aggregates in neurons and chronic neuroinflammation. In the past two decades, in vitro, ex vivo and in vivo studies have consistently shown the involvement of inflammatory responses mediated by microglia and astrocytes, which may be elicited by pathological α-syn or signals from affected neurons and other cell types, and are directly linked to neurodegeneration and disease development. Apart from the prominent immune alterations seen in the CNS, including the infiltration of T cells into the brain, more recent studies have demonstrated important changes in the peripheral immune profile within both the innate and adaptive compartments, particularly involving monocytes, CD4+ and CD8+ T cells. This review aims to integrate the consolidated understanding of immune-related processes underlying the pathogenesis of Parkinson's disease, focusing on both central and peripheral immune cells, neuron-glia crosstalk as well as the central-peripheral immune interaction during the development of Parkinson's disease. Our analysis seeks to provide a comprehensive view of the emerging knowledge of the mechanisms of immunity in Parkinson's disease and the implications of this for better understanding the overall pathogenesis of this disease.
Collapse
Affiliation(s)
- Cintia Roodveldt
- Centre for Molecular Biology and Regenerative Medicine-CABIMER, University of Seville-CSIC, Seville 41092, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville 41009, Spain
| | - Liliana Bernardino
- Health Sciences Research Center (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, 6200-506, Covilhã, Portugal
| | - Ozgur Oztop-Cakmak
- Department of Neurology, Faculty of Medicine, Koç University, Istanbul 34010, Turkey
| | - Milorad Dragic
- Laboratory for Neurobiology, Department of General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
- Department of Molecular Biology and Endocrinology, ‘VINČA’ Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Kari E Fladmark
- Department of Biological Science, University of Bergen, 5006 Bergen, Norway
| | - Sibel Ertan
- Department of Neurology, Faculty of Medicine, Koç University, Istanbul 34010, Turkey
| | - Busra Aktas
- Department of Molecular Biology and Genetics, Burdur Mehmet Akif Ersoy University, Burdur 15200, Turkey
| | - Carlos Pita
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Lucia Ciglar
- Center Health & Bioresources, Competence Unit Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, 1210 Vienna, Austria
| | - Gaetan Garraux
- Movere Group, Faculty of Medicine, GIGA Institute, University of Liège, Liège 4000, Belgium
| | | | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580702, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia 7510156, Santiago, Chile
| | - Marina Romero-Ramos
- Department of Biomedicine & The Danish Research Institute of Translational Neuroscience—DANDRITE, Aarhus University, DK-8000 Aarhus C, Denmark
| |
Collapse
|
4
|
Lin S, Shu Y, Shen R, Zhou Y, Pan H, He L, Fang F, Zhu X, Wang X, Wang Y, Xu W, Ding J. The regulation of NFKB1 on CD200R1 expression and their potential roles in Parkinson's disease. J Neuroinflammation 2024; 21:229. [PMID: 39294682 PMCID: PMC11409543 DOI: 10.1186/s12974-024-03231-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/10/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND Overactivated microglia are a key contributor to Parkinson's disease (PD) by inducing neuroinflammation. CD200R1, a membrane glycoprotein mainly found on microglia, is crucial for maintaining quiescence with its dysregulation linked to microglia's abnormal activation. We and other groups have reported a decline in CD200R1 levels in several neurological disorders including PD. However, the mechanism regulating CD200R1 expression and the specific reasons for its reduction in PD remain largely unexplored. Given the pivotal role of transcription factors in gene expression, this study aimed to elucidate the transcriptional regulation of CD200R1 and its implications in PD. METHODS The CD200R1 promoter core region was identified via luciferase assays. Potential transcription factors were predicted using the UCSC ChIP-seq database and JASPAR. NFKB1 binding to the CD200R1 core promoter was substantiated through electrophoretic mobility shift and chromatin immunoprecipitation assays. Knocking-down or overexpressing NFKB1 validated its regulatory effect on CD200R1. Correlation between decreased CD200R1 and deficient NFKB1 was studied using Genotype-Tissue Expression database. The clinical samples of the peripheral blood mononuclear cells were acquired from 44 PD patients (mean age 64.13 ± 9.78, 43.2% male, median Hoehn-Yahr stage 1.77) and 45 controls (mean age 64.70 ± 9.41, 52.1% male). NFKB1 knockout mice were utilized to study the impact of NFKB1 on CD200R1 expression and to assess their roles in PD pathophysiology. RESULTS The study identified the CD200R1 core promoter region, located 482 to 146 bp upstream of its translation initiation site, was directly regulated by NFKB1. Significant correlation between NFKB1 and CD200R1 expression was observed in human PMBCs. Both NFKB1 and CD200R1 were significantly decreased in PD patient samples. Furthermore, NFKB1-/- mice exhibited exacerbated microglia activation and dopaminergic neuron loss after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment. CONCLUSION Our study identified that NFKB1 served as a direct regulator of CD200R1. Reduced NFKB1 played a critical role in CD200R1 dysregulation and subsequent microglia overactivation in PD. These findings provide evidence that targeting the NFKB1-CD200R1 axis would be a novel therapeutic strategy for PD.
Collapse
Affiliation(s)
- Suzhen Lin
- Department of Neurology, Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yimei Shu
- Department of Neurology, Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ruinan Shen
- Department of Neurology, Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yifan Zhou
- Department of Neurology, Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hong Pan
- Department of Neurology, Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lu He
- Department of Neurology, Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fang Fang
- Department of Aging, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xue Zhu
- Department of Neurology, Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xinrui Wang
- Maternity and child care centers, Fuzhou, Fujian, China
| | - Ying Wang
- Department of Neurology, Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Xu
- Department of Neurology, Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jianqing Ding
- Institute of Aging & Tissue Regeneration, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 160 Pujian Road, Shanghai, 200135, China.
| |
Collapse
|
5
|
Seasons GM, Pellow C, Kuipers HF, Pike GB. Ultrasound and neuroinflammation: immune modulation via the heat shock response. Theranostics 2024; 14:3150-3177. [PMID: 38855178 PMCID: PMC11155413 DOI: 10.7150/thno.96270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024] Open
Abstract
Current pharmacological therapeutic approaches targeting chronic inflammation exhibit transient efficacy, often with adverse effects, limiting their widespread use - especially in the context of neuroinflammation. Effective interventions require the consideration of homeostatic function, pathway dysregulation, and pleiotropic effects when evaluating therapeutic targets. Signalling molecules have multiple functions dependent on the immune context, and this complexity results in therapeutics targeting a single signalling molecule often failing in clinical translation. Additionally, the administration of non-physiologic levels of neurotrophic or anti-inflammatory factors can alter endogenous signalling, resulting in unanticipated effects. Exacerbating these challenges, the central nervous system (CNS) is isolated by the blood brain barrier (BBB), restricting the infiltration of many pharmaceutical compounds into the brain tissue. Consequently, there has been marked interest in therapeutic techniques capable of modulating the immune response in a pleiotropic manner; ultrasound remains on this frontier. While ultrasound has been used therapeutically in peripheral tissues - accelerating healing in wounds, bone fractures, and reducing inflammation - it is only recently that it has been applied to the CNS. The transcranial application of low intensity pulsed ultrasound (LIPUS) has successfully mitigated neuroinflammation in vivo, in models of neurodegenerative disease across a broad spectrum of ultrasound parameters. To date, the underlying biological effects and signalling pathways modulated by ultrasound are poorly understood, with a diverse array of reported molecules implicated. The distributed nature of the beneficial response to LIPUS implies the involvement of an, as yet, undetermined upstream signalling pathway, homologous to the protective effect of febrile range hyperthermia in chronic inflammation. As such, we review the heat shock response (HSR), a protective signalling pathway activated by thermal and mechanical stress, as the possible upstream regulator of the anti-inflammatory effects of ultrasound.
Collapse
Affiliation(s)
- Graham M. Seasons
- Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
| | - Carly Pellow
- Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada
- Department of Radiology, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
| | - Hedwich F. Kuipers
- Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, University of Calgary, Alberta, T2N 1N4, Canada
| | - G. Bruce Pike
- Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
- Department of Radiology, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
| |
Collapse
|
6
|
Augusto-Oliveira M, Tremblay MÈ, Verkhratsky A. Receptors on Microglia. ADVANCES IN NEUROBIOLOGY 2024; 37:83-121. [PMID: 39207688 DOI: 10.1007/978-3-031-55529-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglial cells are the most receptive cells in the central nervous system (CNS), expressing several classes of receptors reflecting their immune heritage and newly acquired neural specialisation. Microglia possess, depending on the particular context, receptors to neurotransmitters and neuromodulators as well as immunocompetent receptors. This rich complement allows microglial cells to monitor the functional status of the nervous system, contribute actively to the regulation of neural activity and plasticity and homeostasis, and guard against pathogens as well as other challenges to the CNS's integrity and function.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
- Programa de Pós-Graduação em Farmacologia e Bioquímica, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Marie-Ève Tremblay
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada
- Department of Molecular Medicine, Université Laval, Pavillon Ferdinand-Vandry, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Life Sciences Center, Vancouver, BC, Canada
| | - Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Manchester, UK.
- Department of Neurosciences, University of the Basque Country, Leioa, Spain.
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
7
|
Morton L, Arndt P, Garza AP, Henneicke S, Mattern H, Gonzalez M, Dityatev A, Yilmazer-Hanke D, Schreiber S, Dunay IR. Spatio-temporal dynamics of microglia phenotype in human and murine cSVD: impact of acute and chronic hypertensive states. Acta Neuropathol Commun 2023; 11:204. [PMID: 38115109 PMCID: PMC10729582 DOI: 10.1186/s40478-023-01672-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/19/2023] [Indexed: 12/21/2023] Open
Abstract
Vascular risk factors such as chronic hypertension are well-established major modifiable factors for the development of cerebral small vessel disease (cSVD). In the present study, our focus was the investigation of cSVD-related phenotypic changes in microglia in human disease and in the spontaneously hypertensive stroke-prone rat (SHRSP) model of cSVD. Our examination of cortical microglia in human post-mortem cSVD cortical tissue revealed distinct morphological microglial features specific to cSVD. We identified enlarged somata, an increase in the territory occupied by thickened microglial processes, and an expansion in the number of vascular-associated microglia. In parallel, we characterized microglia in a rodent model of hypertensive cSVD along different durations of arterial hypertension, i.e., early chronic and late chronic hypertension. Microglial somata were already enlarged in early hypertension. In contrast, at late-stage chronic hypertension, they further exhibited elongated branches, thickened processes, and a reduced ramification index, mirroring the findings in human cSVD. An unbiased multidimensional flow cytometric analysis revealed phenotypic heterogeneity among microglia cells within the hippocampus and cortex. At early-stage hypertension, hippocampal microglia exhibited upregulated CD11b/c, P2Y12R, CD200R, and CD86 surface expression. Detailed analysis of cell subpopulations revealed a unique microglial subset expressing CD11b/c, CD163, and CD86 exclusively in early hypertension. Notably, even at early-stage hypertension, microglia displayed a higher association with cerebral blood vessels. We identified several profound clusters of microglia expressing distinct marker profiles at late chronic hypertensive states. In summary, our findings demonstrate a higher vulnerability of the hippocampus, stage-specific microglial signatures based on morphological features, and cell surface protein expression in response to chronic arterial hypertension. These results indicate the diversity within microglia sub-populations and implicate the subtle involvement of microglia in cSVD pathogenesis.
Collapse
Affiliation(s)
- Lorena Morton
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Health Campus Immunology, Infectiology, and Inflammation (GC-I3), Otto-von-Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Philipp Arndt
- Department of Neurology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) Helmholtz Association, Magdeburg, Germany
| | - Alejandra P Garza
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Health Campus Immunology, Infectiology, and Inflammation (GC-I3), Otto-von-Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Solveig Henneicke
- Department of Neurology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) Helmholtz Association, Magdeburg, Germany
| | - Hendrik Mattern
- German Center for Neurodegenerative Diseases (DZNE) Helmholtz Association, Magdeburg, Germany
- Faculty of Natural Sciences, Biomedical Magnetic Resonance, Otto-von-Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Marilyn Gonzalez
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Health Campus Immunology, Infectiology, and Inflammation (GC-I3), Otto-von-Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE) Helmholtz Association, Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Deniz Yilmazer-Hanke
- Clinical Neuroanatomy, Department of Neurology, Institute for Biomedical Research, Ulm University, Ulm, Germany
| | - Stefanie Schreiber
- Department of Neurology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) Helmholtz Association, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Ildiko R Dunay
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Health Campus Immunology, Infectiology, and Inflammation (GC-I3), Otto-von-Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany.
- Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany.
| |
Collapse
|
8
|
Pfeifer CW, Walsh JT, Santeford A, Lin JB, Beatty WL, Terao R, Liu YA, Hase K, Ruzycki PA, Apte RS. Dysregulated CD200-CD200R signaling in early diabetes modulates microglia-mediated retinopathy. Proc Natl Acad Sci U S A 2023; 120:e2308214120. [PMID: 37903272 PMCID: PMC10636339 DOI: 10.1073/pnas.2308214120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/25/2023] [Indexed: 11/01/2023] Open
Abstract
Diabetic retinopathy (DR) is a neurovascular complication of diabetes. Recent investigations have suggested that early degeneration of the neuroretina may occur prior to the appearance of microvascular changes; however, the mechanisms underlying this neurodegeneration have been elusive. Microglia are the predominant resident immune cell in the retina and adopt dynamic roles in disease. Here, we show that ablation of retinal microglia ameliorates visual dysfunction and neurodegeneration in a type I diabetes mouse model. We also provide evidence of enhanced microglial contact and engulfment of amacrine cells, ultrastructural modifications, and transcriptome changes that drive inflammation and phagocytosis. We show that CD200-CD200R signaling between amacrine cells and microglia is dysregulated during early DR and that targeting CD200R can attenuate high glucose-induced inflammation and phagocytosis in cultured microglia. Last, we demonstrate that targeting CD200R in vivo can prevent visual dysfunction, microglia activation, and retinal inflammation in the diabetic mouse. These studies provide a molecular framework for the pivotal role that microglia play in early DR pathogenesis and identify a potential immunotherapeutic target for treating DR in patients.
Collapse
Affiliation(s)
- Charles W. Pfeifer
- John F. Hardesty, Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO63110
- Neurosciences Graduate Program, Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO63110
| | - James T. Walsh
- John F. Hardesty, Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO63110
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO63110
| | - Andrea Santeford
- John F. Hardesty, Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO63110
| | - Joseph B. Lin
- John F. Hardesty, Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO63110
- Neurosciences Graduate Program, Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO63110
| | - Wandy L. Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO63110
| | - Ryo Terao
- John F. Hardesty, Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO63110
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo1138665, Japan
| | - Yizhou A. Liu
- John F. Hardesty, Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO63110
| | - Keitaro Hase
- John F. Hardesty, Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO63110
| | - Philip A. Ruzycki
- John F. Hardesty, Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO63110
- Department of Genetics, Washington University School of Medicine, St. Louis, MO63110
| | - Rajendra S. Apte
- John F. Hardesty, Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO63110
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO63110
- Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| |
Collapse
|
9
|
Zheng H, Zhang C, Zhang J, Duan L. "Sentinel or accomplice": gut microbiota and microglia crosstalk in disorders of gut-brain interaction. Protein Cell 2023; 14:726-742. [PMID: 37074139 PMCID: PMC10599645 DOI: 10.1093/procel/pwad020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/06/2023] [Indexed: 04/20/2023] Open
Abstract
Abnormal brain-gut interaction is considered the core pathological mechanism behind the disorders of gut-brain interaction (DGBI), in which the intestinal microbiota plays an important role. Microglia are the "sentinels" of the central nervous system (CNS), which participate in tissue damage caused by traumatic brain injury, resist central infection and participate in neurogenesis, and are involved in the occurrence of various neurological diseases. With in-depth research on DGBI, we could find an interaction between the intestinal microbiota and microglia and that they are jointly involved in the occurrence of DGBI, especially in individuals with comorbidities of mental disorders, such as irritable bowel syndrome (IBS). This bidirectional regulation of microbiota and microglia provides a new direction for the treatment of DGBI. In this review, we focus on the role and underlying mechanism of the interaction between gut microbiota and microglia in DGBI, especially IBS, and the corresponding clinical application prospects and highlight its potential to treat DGBI in individuals with psychiatric comorbidities.
Collapse
Affiliation(s)
- Haonan Zheng
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - Cunzheng Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - Jindong Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - Liping Duan
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| |
Collapse
|
10
|
Gao C, Jiang J, Tan Y, Chen S. Microglia in neurodegenerative diseases: mechanism and potential therapeutic targets. Signal Transduct Target Ther 2023; 8:359. [PMID: 37735487 PMCID: PMC10514343 DOI: 10.1038/s41392-023-01588-0] [Citation(s) in RCA: 183] [Impact Index Per Article: 91.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/11/2023] [Accepted: 08/03/2023] [Indexed: 09/23/2023] Open
Abstract
Microglia activation is observed in various neurodegenerative diseases. Recent advances in single-cell technologies have revealed that these reactive microglia were with high spatial and temporal heterogeneity. Some identified microglia in specific states correlate with pathological hallmarks and are associated with specific functions. Microglia both exert protective function by phagocytosing and clearing pathological protein aggregates and play detrimental roles due to excessive uptake of protein aggregates, which would lead to microglial phagocytic ability impairment, neuroinflammation, and eventually neurodegeneration. In addition, peripheral immune cells infiltration shapes microglia into a pro-inflammatory phenotype and accelerates disease progression. Microglia also act as a mobile vehicle to propagate protein aggregates. Extracellular vesicles released from microglia and autophagy impairment in microglia all contribute to pathological progression and neurodegeneration. Thus, enhancing microglial phagocytosis, reducing microglial-mediated neuroinflammation, inhibiting microglial exosome synthesis and secretion, and promoting microglial conversion into a protective phenotype are considered to be promising strategies for the therapy of neurodegenerative diseases. Here we comprehensively review the biology of microglia and the roles of microglia in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, multiple system atrophy, amyotrophic lateral sclerosis, frontotemporal dementia, progressive supranuclear palsy, corticobasal degeneration, dementia with Lewy bodies and Huntington's disease. We also summarize the possible microglia-targeted interventions and treatments against neurodegenerative diseases with preclinical and clinical evidence in cell experiments, animal studies, and clinical trials.
Collapse
Affiliation(s)
- Chao Gao
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jingwen Jiang
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yuyan Tan
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Shengdi Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, 201210, Shanghai, China.
| |
Collapse
|
11
|
Chen X, Cui QQ, Hu XH, Ye J, Liu ZC, Mei YX, Wang F, Hu ZL, Chen JG. CD200 in dentate gyrus improves depressive-like behaviors of mice through enhancing hippocampal neurogenesis via alleviation of microglia hyperactivation. J Neuroinflammation 2023; 20:157. [PMID: 37391731 DOI: 10.1186/s12974-023-02836-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/16/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND Neuroinflammation and microglia play critical roles in the development of depression. Cluster of differentiation 200 (CD200) is an anti-inflammatory glycoprotein that is mainly expressed in neurons, and its receptor CD200R1 is primarily in microglia. Although the CD200-CD200R1 pathway is necessary for microglial activation, its role in the pathophysiology of depression remains unknown. METHODS The chronic social defeat stress (CSDS) with behavioral tests were performed to investigate the effect of CD200 on the depressive-like behaviors. Viral vectors were used to overexpress or knockdown of CD200. The levels of CD200 and inflammatory cytokines were tested with molecular biological techniques. The status of microglia, the expression of BDNF and neurogenesis were detected with immunofluorescence imaging. RESULTS We found that the expression of CD200 was decreased in the dentate gyrus (DG) region of mice experienced CSDS. Overexpression of CD200 alleviated the depressive-like behaviors of stressed mice and inhibition of CD200 facilitated the susceptibility to stress. When CD200R1 receptors on microglia were knocked down, CD200 was unable to exert its role in alleviating depressive-like behavior. Microglia in the DG brain region were morphologically activated after exposure to CSDS. In contrast, exogenous administration of CD200 inhibited microglia hyperactivation, alleviated neuroinflammatory response in hippocampus, and increased the expression of BDNF, which in turn ameliorated adult hippocampal neurogenesis impairment in the DG induced by CSDS. CONCLUSIONS Taken together, these results suggest that CD200-mediated alleviation of microglia hyperactivation contributes to the antidepressant effect of neurogenesis in dentate gyrus in mice.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian-Qian Cui
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Hai Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Ye
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zi-Cun Liu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan-Xi Mei
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
- The Research Center for Depression, Tongji Medical College, Huazhong University of Science, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China
| | - Zhuang-Li Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China.
- The Research Center for Depression, Tongji Medical College, Huazhong University of Science, Wuhan, 430030, China.
- Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China.
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China.
- The Research Center for Depression, Tongji Medical College, Huazhong University of Science, Wuhan, 430030, China.
- Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China.
| |
Collapse
|
12
|
Strader S, West AB. The interplay between monocytes, α-synuclein and LRRK2 in Parkinson's disease. Biochem Soc Trans 2023; 51:747-758. [PMID: 37013975 PMCID: PMC11110874 DOI: 10.1042/bst20201091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 04/05/2023]
Abstract
The accumulation of aggregated α-synuclein in susceptible neurons in the brain, together with robust activation of nearby myeloid cells, are pathological hallmarks of Parkinson's disease (PD). While microglia represent the dominant type of myeloid cell in the brain, recent genetic and whole-transcriptomic studies have implicated another type of myeloid cell, bone-marrow derived monocytes, in disease risk and progression. Monocytes in circulation harbor high concentrations of the PD-linked enzyme leucine-rich repeat kinase 2 (LRRK2) and respond to both intracellular and extracellular aggregated α-synuclein with a variety of strong pro-inflammatory responses. This review highlights recent findings from studies that functionally characterize monocytes in PD patients, monocytes that infiltrate into cerebrospinal fluid, and emerging analyses of whole myeloid cell populations in the PD-affected brain that include monocyte populations. Central controversies discussed include the relative contribution of monocytes acting in the periphery from those that might engraft in the brain to modify disease risk and progression. We conclude that further investigation into monocyte pathways and responses in PD, especially the discovery of additional markers, transcriptomic signatures, and functional classifications, that better distinguish monocyte lineages and responses in the brain from other types of myeloid cells may reveal points for therapeutic intervention, as well as a better understanding of ongoing inflammation associated with PD.
Collapse
Affiliation(s)
- Samuel Strader
- Duke Center for Neurodegeneration and Neurotherapeutics, Department of Pharmacology and Cancer Biology, Duke University, 3 Genome Court, Durham, 27710, North Carolina, U.S.A
| | - Andrew B. West
- Duke Center for Neurodegeneration and Neurotherapeutics, Department of Pharmacology and Cancer Biology, Duke University, 3 Genome Court, Durham, 27710, North Carolina, U.S.A
| |
Collapse
|
13
|
Microglial Activation in Metal Neurotoxicity: Impact in Neurodegenerative Diseases. BIOMED RESEARCH INTERNATIONAL 2023; 2023:7389508. [PMID: 36760476 PMCID: PMC9904912 DOI: 10.1155/2023/7389508] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 02/04/2023]
Abstract
Neurodegenerative processes encompass a large variety of diseases with different pathological patterns and clinical features, such as Alzheimer's and Parkinson's diseases. Exposure to metals has been hypothesized to increase oxidative stress in brain cells leading to cell death and neurodegeneration. Neurotoxicity of metals has been demonstrated by several in vitro and in vivo experimental studies, and most probably, each metal has its specific pathway to trigger cell death. As a result, exposure to essential metals, such as manganese, iron, copper, zinc, and cobalt, and nonessential metals, including lead, aluminum, and cadmium, perturbs metal homeostasis at the cellular and organism levels leading to neurodegeneration. In this contribution, a comprehensive review of the molecular mechanisms by which metals affect microglia physiology and signaling properties is presented. Furthermore, studies that validate the disruption of microglia activation pathways as an essential mechanism of metal toxicity that can contribute to neurodegenerative disease are also presented and discussed.
Collapse
|
14
|
Rangan P, Lobo F, Parrella E, Rochette N, Morselli M, Stephen TL, Cremonini AL, Tagliafico L, Persia A, Caffa I, Monacelli F, Odetti P, Bonfiglio T, Nencioni A, Pigliautile M, Boccardi V, Mecocci P, Pike CJ, Cohen P, LaDu MJ, Pellegrini M, Xia K, Tran K, Ann B, Chowdhury D, Longo VD. Fasting-mimicking diet cycles reduce neuroinflammation to attenuate cognitive decline in Alzheimer's models. Cell Rep 2022; 40:111417. [PMID: 36170815 PMCID: PMC9648488 DOI: 10.1016/j.celrep.2022.111417] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/30/2022] [Accepted: 09/01/2022] [Indexed: 11/22/2022] Open
Abstract
The effects of fasting-mimicking diet (FMD) cycles in reducing many aging and disease risk factors indicate it could affect Alzheimer's disease (AD). Here, we show that FMD cycles reduce cognitive decline and AD pathology in E4FAD and 3xTg AD mouse models, with effects superior to those caused by protein restriction cycles. In 3xTg mice, long-term FMD cycles reduce hippocampal Aβ load and hyperphosphorylated tau, enhance genesis of neural stem cells, decrease microglia number, and reduce expression of neuroinflammatory genes, including superoxide-generating NADPH oxidase (Nox2). 3xTg mice lacking Nox2 or mice treated with the NADPH oxidase inhibitor apocynin also display improved cognition and reduced microglia activation compared with controls. Clinical data indicate that FMD cycles are feasible and generally safe in a small group of AD patients. These results indicate that FMD cycles delay cognitive decline in AD models in part by reducing neuroinflammation and/or superoxide production in the brain.
Collapse
Affiliation(s)
- Priya Rangan
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Fleur Lobo
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Edoardo Parrella
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA; Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, BS 25123, Italy
| | - Nicolas Rochette
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 611 Charles E. Young Dr. E., Los Angeles, CA 90095, USA; Department of Ecology and Evolutionary Biology, University of California, Los Angeles, 612 Charles E. Young Dr. E., Los Angeles, CA 90095, USA
| | - Marco Morselli
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 610 Charles E. Young Dr. S., Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 611 Charles E. Young Dr. E., Los Angeles, CA 90095, USA
| | - Terri-Leigh Stephen
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Anna Laura Cremonini
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Luca Tagliafico
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy
| | - Angelica Persia
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy
| | - Irene Caffa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Patrizio Odetti
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Tommaso Bonfiglio
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Martina Pigliautile
- Santa Maria della Misericordia Hospital, Section of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Virginia Boccardi
- Santa Maria della Misericordia Hospital, Section of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Patrizia Mecocci
- Santa Maria della Misericordia Hospital, Section of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Christian J Pike
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Pinchas Cohen
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA; USC Dornsife College of Letters, Arts & Sciences, Department of Biological Sciences, University of Southern California, 3551 Trousdale Pkwy., Los Angeles, CA 90089-0191, USA
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 610 Charles E. Young Dr. S., Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 611 Charles E. Young Dr. E., Los Angeles, CA 90095, USA
| | - Kyle Xia
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Katelynn Tran
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Brandon Ann
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Dolly Chowdhury
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Valter D Longo
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA; Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, 1425 San Pablo St., Los Angeles, CA 90033, USA; IFOM FIRC Institute of Molecular Oncology, Via Adamello 16, Milano, MI 20139, Italy.
| |
Collapse
|
15
|
Zhang F, Ran Y, Tahir M, Li Z, Wang J, Chen X. Regulation of N6-methyladenosine (m6A) RNA methylation in microglia-mediated inflammation and ischemic stroke. Front Cell Neurosci 2022; 16:955222. [PMID: 35990887 PMCID: PMC9386152 DOI: 10.3389/fncel.2022.955222] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
N6-methyladenosine (m6A) is the most abundant post-transcription modification, widely occurring in eukaryotic mRNA and non-coding RNA. m6A modification is highly enriched in the mammalian brain and is associated with neurological diseases like Alzheimer’s disease (AD) and Parkinson’s disease (PD). Ischemic stroke (IS) was discovered to alter the cerebral m6A epi-transcriptome, which might have functional implications in post-stroke pathophysiology. Moreover, it is observed that m6A modification could regulate microglia’s pro-inflammatory and anti-inflammatory responses. Given the critical regulatory role of microglia in the inflammatory processes in the central nervous system (CNS), we speculate that m6A modification could modulate the post-stroke microglial inflammatory responses. This review summarizes the vital regulatory roles of m6A modification in microglia-mediated inflammation and IS. Stroke is associated with a high recurrence rate, understanding the relationship between m6A modification and stroke may help stroke rehabilitation and develop novel therapies in the future.
Collapse
Affiliation(s)
- Fangfang Zhang
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Yuanyuan Ran
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Muhammad Tahir
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Zihan Li
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Jianan Wang
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Xuechai Chen
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Faculty of Environment and Life, Beijing University of Technology, Beijing, China
- *Correspondence: Xuechai Chen,
| |
Collapse
|
16
|
Jia Y, Yao Y, Zhuo L, Chen X, Yan C, Ji Y, Tao J, Zhu Y. Aerobic Physical Exercise as a Non-medical Intervention for Brain Dysfunction: State of the Art and Beyond. Front Neurol 2022; 13:862078. [PMID: 35645958 PMCID: PMC9136296 DOI: 10.3389/fneur.2022.862078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/28/2022] [Indexed: 12/03/2022] Open
Abstract
Brain disorders, including stroke, Alzheimer's disease, depression, and chronic pain, are difficult to effectively treat. These major brain disorders have high incidence and mortality rates in the general population, and seriously affect not only the patient's quality of life, but also increases the burden of social medical care. Aerobic physical exercise is considered an effective adjuvant therapy for preventing and treating major brain disorders. Although the underlying regulatory mechanisms are still unknown, systemic processes may be involved. Here, this review aimed to reveal that aerobic physical exercise improved depression and several brain functions, including cognitive functions, and provided chronic pain relief. We concluded that aerobic physical exercise helps to maintain the regulatory mechanisms of brain homeostasis through anti-inflammatory mechanisms and enhanced synaptic plasticity and inhibition of hippocampal atrophy and neuronal apoptosis. In addition, we also discussed the cross-system mechanisms of aerobic exercise in regulating imbalances in brain function, such as the “bone-brain axis.” Furthermore, our findings provide a scientific basis for the clinical application of aerobic physical exercise in the fight against brain disorders.
Collapse
Affiliation(s)
- Yuxiang Jia
- School of Medicine and School of Life Sciences, Shanghai University, Shanghai, China
| | - Yu Yao
- School of Medicine and School of Life Sciences, Shanghai University, Shanghai, China
| | - Limin Zhuo
- School of Medicine and School of Life Sciences, Shanghai University, Shanghai, China
| | - Xingxing Chen
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cuina Yan
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yonghua Ji
- School of Medicine and School of Life Sciences, Shanghai University, Shanghai, China
- *Correspondence: Yonghua Ji
| | - Jie Tao
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Jie Tao
| | - Yudan Zhu
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Yudan Zhu
| |
Collapse
|
17
|
Hiraga SI, Itokazu T, Nishibe M, Yamashita T. Neuroplasticity related to chronic pain and its modulation by microglia. Inflamm Regen 2022; 42:15. [PMID: 35501933 PMCID: PMC9063368 DOI: 10.1186/s41232-022-00199-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/19/2022] [Indexed: 01/03/2023] Open
Abstract
Neuropathic pain is often chronic and can persist after overt tissue damage heals, suggesting that its underlying mechanism involves the alteration of neuronal function. Such an alteration can be a direct consequence of nerve damage or a result of neuroplasticity secondary to the damage to tissues or to neurons. Recent studies have shown that neuroplasticity is linked to causing neuropathic pain in response to nerve damage, which may occur adjacent to or remotely from the site of injury. Furthermore, studies have revealed that neuroplasticity relevant to chronic pain is modulated by microglia, resident immune cells of the central nervous system (CNS). Microglia may directly contribute to synaptic remodeling and altering pain circuits, or indirectly contribute to neuroplasticity through property changes, including the secretion of growth factors. We herein highlight the mechanisms underlying neuroplasticity that occur in the somatosensory circuit of the spinal dorsal horn, thalamus, and cortex associated with chronic pain following injury to the peripheral nervous system (PNS) or CNS. We also discuss the dynamic functions of microglia in shaping neuroplasticity related to chronic pain. We suggest further understanding of post-injury ectopic plasticity in the somatosensory circuits may shed light on the differential mechanisms underlying nociceptive, neuropathic, and nociplastic-type pain. While one of the prominent roles played by microglia appears to be the modulation of post-injury neuroplasticity. Therefore, future molecular- or genetics-based studies that address microglia-mediated post-injury neuroplasticity may contribute to the development of novel therapies for chronic pain.
Collapse
Affiliation(s)
- Shin-Ichiro Hiraga
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takahide Itokazu
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan. .,Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Mariko Nishibe
- Center for Strategic Innovative Dentistry, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| | - Toshihide Yamashita
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan. .,Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan. .,WPI Immunology Frontier Research Center, Osaka, Japan. .,Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
| |
Collapse
|
18
|
Callai EMM, Zin LEF, Catarina LS, Ponzoni D, Gonçalves CAS, Vizuete AFK, Cougo MC, Boff J, Puricelli E, Fernandes EK, da Silva Torres IL, Quevedo AS. Evaluation of the immediate effects of a single transcranial direct current stimulation session on astrocyte activation, inflammatory response, and pain threshold in naïve rats. Behav Brain Res 2022; 428:113880. [PMID: 35390432 DOI: 10.1016/j.bbr.2022.113880] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 02/15/2022] [Accepted: 04/01/2022] [Indexed: 11/18/2022]
Abstract
Transcranial direct current stimulation (tDCS) has demonstrated clinical benefits such as analgesia, anti-inflammatory, and neuroprotective effects. However, the mechanisms of action of a single tDCS session are poorly characterized. The present study aimed to evaluate the effects of a single tDCS session on pain sensitivity, inflammatory parameters, and astrocyte activity in naive rats. In the first experiment, sixty-day-old male Wistar rats (n=95) were tested for mechanical pain threshold (von Frey test). Afterward, animals were submitted to a single bimodal tDCS (0.5mA, 20minutes) or sham-tDCS session. According to the group, animals were re-tested at different time intervals (30, 60, 120minutes, or 24hours) after the intervention, euthanized, and the cerebral cortex collected for biochemical analysis. A second experiment (n=16) was performed using a similar protocol to test the hypotheses that S100B levels in the cerebrospinal fluid (CSF) are altered by tDCS. Elisa assay quantified the levels of tumor necrosis factor-alfa (TNF-α), interleukin-10 (IL10), S100 calcium-binding protein B (S100B), and Glial fibrillary acidic protein (GFAP). Data were analyzed using ANOVA and independent t-test (P<0.05). Results showed that tDCS decreased pain sensitivity (30 and 60min), cerebral TNF-α and S100B levels (30min). CSF S100B levels increased 30minutes after intervention. There were no differences in IL10 and GFAP levels. TCDS showed analgesic, anti-inflammatory, and neuroprotective effects in naive animals. Therefore, this non-invasive and inexpensive therapy may potentially be a preemptive alternative to reduce pain, inflammation, and neurodegeneration in situations where patients will undergo medical procedures (e.g., surgery).
Collapse
Affiliation(s)
- Etiane Micheli Meyer Callai
- Postgraduate Program in Dentistry, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, RS, Brazil
| | | | - Luciana Santa Catarina
- Postgraduate Program in Dentistry, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, RS, Brazil
| | - Deise Ponzoni
- Postgraduate Program in Dentistry, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, RS, Brazil
| | | | | | - Milton Cristian Cougo
- Postgraduate Program in Dentistry, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, RS, Brazil
| | - Jamile Boff
- Postgraduate Program in Dentistry, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, RS, Brazil
| | - Edela Puricelli
- Postgraduate Program in Dentistry, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, RS, Brazil
| | | | | | - Alexandre Silva Quevedo
- Postgraduate Program in Dentistry, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, RS, Brazil; Neuroscience Graduate Program, UFRGS, Porto Alegre, RS, Brazil; Pharmacology Graduate Program, UFRGS, Porto Alegre, RS, Brazil.
| |
Collapse
|
19
|
Altered expression of the immunoregulatory ligand-receptor pair CD200-CD200R1 in the brain of Parkinson's disease patients. NPJ Parkinsons Dis 2022; 8:27. [PMID: 35296683 PMCID: PMC8927151 DOI: 10.1038/s41531-022-00290-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 02/17/2022] [Indexed: 12/22/2022] Open
Abstract
Neuroinflammation, in which activated microglia are involved, appears to contribute to the development of Parkinson’s disease (PD). However, the role of microglial activation and the mechanisms governing this process remain uncertain. We focused on one inhibitory mechanism involved in the control of microglial activation, the microglia inhibitory receptor CD200R1, and its ligand CD200, mainly expressed by neurons. The human CD200R1 gene encodes two membrane-associated and two soluble protein isoforms and the human CD200 gene encodes full-length proteins (CD200full) but also truncated (CD200tr) proteins which act as CD200R1 antagonists. Little is known about their expression in the human brain under pathological conditions. We used human peripheral blood monocytes and monocyte-derived microglia-like cells from control subjects to characterize the expression of the CD200R1 mRNA variants, which showed stimulus-specific responses. We provide evidence of increased CD200R1 (mRNA variants and protein isoforms) and CD200 expression (CD200tr mRNA) in brain tissue of PD patients, mainly in the hippocampus, as well as increased CD200 expression (CD200full and CD200tr mRNAs) in iPSCs-derived dopaminergic neurons generated from skin fibroblasts of PD patients. Our results suggest that CD200-CD200R1 signalling is altered in PD, which may affect the microglial function and constitute a potential target in therapeutic strategies for PD.
Collapse
|
20
|
Abstract
The notion that autoimmune responses to α-synuclein may be involved in the pathogenesis of this disorder stems from reports that mutations in α-synuclein or certain alleles of the major histocompatibility complex (MHC) are associated with the disease and that dopaminergic and norepinephrinergic neurons in the midbrain can present antigenic epitopes. Here, we discuss recent evidence that a defined set of peptides derived from α-synuclein act as antigenic epitopes displayed by specific MHC alleles and drive helper and cytotoxic T cell responses in patients with PD. Moreover, phosphorylated α-synuclein may activate T cell responses in a less restricted manner in PD. While the roles for the acquired immune system in disease pathogenesis remain unknown, preclinical animal models and in vitro studies indicate that T cells may interact with neurons and exert effects related to neuronal death and neuroprotection. These findings suggest that therapeutics that target T cells and ameliorate the incidence or disease severity of inflammatory bowel disorders or CNS autoimmune diseases such as multiple sclerosis may be useful in PD.
Collapse
|
21
|
Singh V, Kushwaha S, Ansari JA, Gangopadhyay S, Mishra SK, Dey RK, Giri AK, Patnaik S, Ghosh D. MicroRNA-129-5p-regulated microglial expression of the surface receptor CD200R1 controls neuroinflammation. J Biol Chem 2021; 298:101521. [PMID: 34952004 PMCID: PMC8762073 DOI: 10.1016/j.jbc.2021.101521] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 11/28/2022] Open
Abstract
CD200R1 is an inhibitory surface receptor expressed in microglia and blood macrophages. Microglial CD200R1 is known to control neuroinflammation by keeping the microglia in resting state, and therefore, tight regulation of its expression is important. CCAAT/enhancer-binding protein β (CEBPβ) is the known regulator of CD200R1 transcription. In the present study, our specific intention was to find a possible posttranscriptional regulatory mechanism of CD200R1 expression. Here we investigated a novel regulatory mechanism of CD200R1 expression following exposure to an environmental stressor, arsenic, combining in silico analysis, in vitro, and in vivo experiments, as well as validation in human samples. The in silico analysis and in vitro studies with primary neonatal microglia and BV2 microglia revealed that arsenic demethylates the promoter of a microRNA, miR-129-5p, thereby increasing its expression, which subsequently represses CD200R1 by binding to its 3′-untranslated region and shuttling the CD200R1 mRNA to the cytoplasmic-processing body in mouse microglia. The role of miR-129-5p was further validated in BALB/c mouse by stereotaxically injecting anti-miR-129. We found that anti-miR-129 reversed the expression of CD200R1, as well as levels of inflammatory molecules IL-6 and TNF-α. Experiments with a CD200R1 siRNA-induced loss-of-function mouse model confirmed an miR-129-5p→CD200R1→IL-6/TNF-α signaling axis. These main findings were replicated in a human cell line and validated in human samples. Taken together, our study revealed miR-129-5p as a novel posttranscriptional regulator of CD200R1 expression with potential implications in neuroinflammation and related complications.
Collapse
Affiliation(s)
- Vikas Singh
- Immunotoxicology Laboratory, Food, Drug & Chemical Toxicology Group and Nanomaterial Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shaivya Kushwaha
- Immunotoxicology Laboratory, Food, Drug & Chemical Toxicology Group and Nanomaterial Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Jamal Ahmad Ansari
- Immunotoxicology Laboratory, Food, Drug & Chemical Toxicology Group and Nanomaterial Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Siddhartha Gangopadhyay
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Developmental Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India
| | - Shubhendra K Mishra
- Immunotoxicology Laboratory, Food, Drug & Chemical Toxicology Group and Nanomaterial Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India
| | - Rajib K Dey
- Immunotoxicology Laboratory, Food, Drug & Chemical Toxicology Group and Nanomaterial Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ashok K Giri
- CSIR-Indian Institute of Chemical Biology, 4, Raja Subodh Chandra Mallick Rd, Poddar Nagar, Jadavpur, Kolkata, West Bengal 700032, India
| | - Satyakam Patnaik
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Water Analysis Laboratory, Nanomaterial Toxicology Group, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh 226001, India
| | - Debabrata Ghosh
- Immunotoxicology Laboratory, Food, Drug & Chemical Toxicology Group and Nanomaterial Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
22
|
Macrophages transfer mitochondria to sensory neurons to resolve inflammatory pain. Neuron 2021; 110:613-626.e9. [PMID: 34921782 DOI: 10.1016/j.neuron.2021.11.020] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/21/2021] [Accepted: 11/16/2021] [Indexed: 12/21/2022]
Abstract
The current paradigm is that inflammatory pain passively resolves following the cessation of inflammation. Yet, in a substantial proportion of patients with inflammatory diseases, resolution of inflammation is not sufficient to resolve pain, resulting in chronic pain. Mechanistic insight into how inflammatory pain is resolved is lacking. Here, we show that macrophages actively control resolution of inflammatory pain remotely from the site of inflammation by transferring mitochondria to sensory neurons. During resolution of inflammatory pain in mice, M2-like macrophages infiltrate the dorsal root ganglia that contain the somata of sensory neurons, concurrent with the recovery of oxidative phosphorylation in sensory neurons. The resolution of pain and the transfer of mitochondria requires expression of CD200 receptor (CD200R) on macrophages and the non-canonical CD200R-ligand iSec1 on sensory neurons. Our data reveal a novel mechanism for active resolution of inflammatory pain.
Collapse
|
23
|
Feige L, Zaeck LM, Sehl-Ewert J, Finke S, Bourhy H. Innate Immune Signaling and Role of Glial Cells in Herpes Simplex Virus- and Rabies Virus-Induced Encephalitis. Viruses 2021; 13:2364. [PMID: 34960633 PMCID: PMC8708193 DOI: 10.3390/v13122364] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 12/19/2022] Open
Abstract
The environment of the central nervous system (CNS) represents a double-edged sword in the context of viral infections. On the one hand, the infectious route for viral pathogens is restricted via neuroprotective barriers; on the other hand, viruses benefit from the immunologically quiescent neural environment after CNS entry. Both the herpes simplex virus (HSV) and the rabies virus (RABV) bypass the neuroprotective blood-brain barrier (BBB) and successfully enter the CNS parenchyma via nerve endings. Despite the differences in the molecular nature of both viruses, each virus uses retrograde transport along peripheral nerves to reach the human CNS. Once inside the CNS parenchyma, HSV infection results in severe acute inflammation, necrosis, and hemorrhaging, while RABV preserves the intact neuronal network by inhibiting apoptosis and limiting inflammation. During RABV neuroinvasion, surveilling glial cells fail to generate a sufficient type I interferon (IFN) response, enabling RABV to replicate undetected, ultimately leading to its fatal outcome. To date, we do not fully understand the molecular mechanisms underlying the activation or suppression of the host inflammatory responses of surveilling glial cells, which present important pathways shaping viral pathogenesis and clinical outcome in viral encephalitis. Here, we compare the innate immune responses of glial cells in RABV- and HSV-infected CNS, highlighting different viral strategies of neuroprotection or Neuroinflamm. in the context of viral encephalitis.
Collapse
Affiliation(s)
- Lena Feige
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology, 28 Rue Du Docteur Roux, 75015 Paris, France;
| | - Luca M. Zaeck
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut (FLI), Federal Institute of Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany; (L.M.Z.); (S.F.)
| | - Julia Sehl-Ewert
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut (FLI), Federal Institute of Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany;
| | - Stefan Finke
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut (FLI), Federal Institute of Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany; (L.M.Z.); (S.F.)
| | - Hervé Bourhy
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology, 28 Rue Du Docteur Roux, 75015 Paris, France;
| |
Collapse
|
24
|
Al Mamun A, Ngwa C, Qi S, Honarpisheh P, Datar S, Sharmeen R, Xu Y, McCullough LD, Liu F. Neuronal CD200 Signaling Is Protective in the Acute Phase of Ischemic Stroke. Stroke 2021; 52:3362-3373. [PMID: 34353112 DOI: 10.1161/strokeaha.120.032374] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE CD200 (cluster of differentiation 200), a highly glycosylated protein primarily expressed on neurons in the central nervous system, binds with its receptor CD200R to form an endogenous inhibitory signal against immune responses. However, little is known about the effect of neuronal CD200 signaling in cerebral ischemia. The aim of this study was to investigate how neuronal CD200 signaling impacts poststroke inflammation and the ischemic injury. METHODS CD200 tma1lf/fl:Thy1CreER mice were treated with tamoxifen to induce conditional gene knockout (ICKO) of neuronal CD200. The mice were subjected to a 60-minute transient middle cerebral artery occlusion. Stroke outcomes, apoptotic cell death, immune cell infiltration, microglia activation, and other inflammatory profiles were evaluated at 3 and 7 days after stroke. RESULTS Infarct volumes were significantly larger, and behavioral deficits more severe in ICKO versus control mice at 3 days after middle cerebral artery occlusion. Terminal deoxynucleotidyl transferase dUTP nick end labeling assay also revealed a significant increase in apoptotic neuronal death in CD200 ICKO mice. An enhancement in lymphocytic infiltration and microglial proinflammatory responses were revealed by flow cytometry at 3 and 7 days after stroke in ICKO mice, accompanied by an increased microglial phagocytosis activity. Plasma proinflammatory cytokine (TNFα [tumor necrosis factor alpha] and IL [interleukin]-1β) levels significantly increased at 3 days, and IL-1β/IL-6 levels increased at 7 days in ICKO versus control animals. ICKO led to significantly lower baseline level of CD200 both in brain and plasma. CONCLUSIONS Neuronal CD200 inhibits proinflammatory responses and is protective against stroke injury.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Conelius Ngwa
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Shaohua Qi
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Pedram Honarpisheh
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Saumil Datar
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Romana Sharmeen
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Yan Xu
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Louise D McCullough
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Fudong Liu
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| |
Collapse
|
25
|
Semenova E, Grudniak MP, Machaj EK, Bocian K, Chroscinska-Krawczyk M, Trochonowicz M, Stepaniec IM, Murzyn M, Zagorska KE, Boruczkowski D, Kolanowski TJ, Oldak T, Rozwadowska N. Mesenchymal Stromal Cells from Different Parts of Umbilical Cord: Approach to Comparison & Characteristics. Stem Cell Rev Rep 2021; 17:1780-1795. [PMID: 33860454 PMCID: PMC8553697 DOI: 10.1007/s12015-021-10157-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2021] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal/stem cells (MSCs) are a unique population of cells that play an important role in the regeneration potential of the body. MSCs exhibit a characteristic phenotype and are capable of modulating the immune response. MSCs can be isolated from various tissues such as: bone marrow, adipose tissue, placenta, umbilical cord and others. The umbilical cord as a source of MSCs, has strong advantages, such as no-risk procedure of tissue retrieval after birth and easiness of the MSCs isolation. As the umbilical cord (UC) is a complex organ and we decided to evaluate, whether the cells derived from different regions of umbilical cord show similar or distinct properties. In this study we characterized and compared MSCs from three regions of the umbilical cord: Wharton's Jelly (WJ), the perivascular space (PRV) and the umbilical membrane (UCM). The analysis was carried out in terms of morphology, phenotype, immunomodulation potential and secretome. Based on the obtained results, we were able to conclude, that MSCs derived from distinct UC regions differ in their properties. According to our result WJ-MSCs have high and stabile proliferation potential and phenotype, when compare with other MSCs and can be treated as a preferable source of cells for medical application.
Collapse
Affiliation(s)
- Ekaterina Semenova
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Mariusz P Grudniak
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Eugeniusz K Machaj
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Katarzyna Bocian
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland.,Faculty of Biology, Department of Immunology, University of Warsaw, Warsaw, Poland
| | | | - Marzena Trochonowicz
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Igor M Stepaniec
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Magdalena Murzyn
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland.,Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Karolina E Zagorska
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Dariusz Boruczkowski
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Tomasz J Kolanowski
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland.,Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Tomasz Oldak
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland.
| | | |
Collapse
|
26
|
Rabaneda-Lombarte N, Serratosa J, Bové J, Vila M, Saura J, Solà C. The CD200R1 microglial inhibitory receptor as a therapeutic target in the MPTP model of Parkinson's disease. J Neuroinflammation 2021; 18:88. [PMID: 33823877 PMCID: PMC8025338 DOI: 10.1186/s12974-021-02132-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 03/15/2021] [Indexed: 01/22/2023] Open
Abstract
Background It is suggested that neuroinflammation, in which activated microglial cells play a relevant role, contributes to the development of Parkinson’s disease (PD). Consequently, the modulation of microglial activation is a potential therapeutic target to be taken into account to act against the dopaminergic neurodegeneration occurring in this neurological disorder. Several soluble and membrane-associated inhibitory mechanisms contribute to maintaining microglial cells in a quiescent/surveillant phenotype in physiological conditions. However, the presence of activated microglial cells in the brain in PD patients suggests that these mechanisms have been somehow overloaded. We focused our interest on one of the membrane-associated mechanisms, the CD200-CD200R1 ligand-receptor pair. Methods The acute MPTP experimental mouse model of PD was used to study the temporal pattern of mRNA expression of CD200 and CD200R1 in the context of MPTP-induced dopaminergic neurodegeneration and neuroinflammation. Dopaminergic damage was assessed by tyrosine hydroxylase (TH) immunoreactivity, and neuroinflammation was evaluated by the mRNA expression of inflammatory markers and IBA1 and GFAP immunohistochemistry. The effect of the modulation of the CD200-CD200R1 system on MPTP-induced damage was determined by using a CD200R1 agonist or CD200 KO mice. Results MPTP administration resulted in a progressive decrease in TH-positive fibres in the striatum and TH-positive neurons in the substantia nigra pars compacta, which were accompanied by transient astrogliosis, microgliosis and expression of pro- and anti-inflammatory markers. CD200 mRNA levels rapidly decreased in the ventral midbrain after MPTP treatment, while a transient decrease of CD200R1 mRNA expression was repeatedly observed in this brain area at earlier and later phases. By contrast, a transient increase in CD200R1 expression was observed in striatum. The administration of a CD200R1 agonist resulted in the inhibition of MPTP-induced dopaminergic neurodegeneration, while microglial cells showed signs of earlier activation in CD200-deficient mice. Conclusions Collectively, these findings provide evidence for a correlation between CD200-CD200R1 alterations, glial activation and neuronal loss. CD200R1 stimulation reduces MPTP-induced loss of dopaminergic neurons, and CD200 deficiency results in earlier microglial activation, suggesting that the potentiation of CD200R1 signalling is a possible approach to controlling neuroinflammation and neuronal death in PD.
Collapse
Affiliation(s)
- Neus Rabaneda-Lombarte
- Department of Cerebral Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August-Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Biochemistry and Molecular Biology Unit, School of Medicine, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Joan Serratosa
- Department of Cerebral Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August-Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jordi Bové
- Vall d'Hebrón Research Institute-CIBERNED, Barcelona, Spain
| | - Miquel Vila
- Vall d'Hebrón Research Institute-CIBERNED, Barcelona, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Josep Saura
- Biochemistry and Molecular Biology Unit, School of Medicine, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Carme Solà
- Department of Cerebral Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August-Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
27
|
Bogale TA, Faustini G, Longhena F, Mitola S, Pizzi M, Bellucci A. Alpha-Synuclein in the Regulation of Brain Endothelial and Perivascular Cells: Gaps and Future Perspectives. Front Immunol 2021; 12:611761. [PMID: 33679750 PMCID: PMC7933041 DOI: 10.3389/fimmu.2021.611761] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/27/2021] [Indexed: 12/27/2022] Open
Abstract
Misfolded proteins, inflammation, and vascular alterations are common pathological hallmarks of neurodegenerative diseases. Alpha-synuclein is a small synaptic protein that was identified as a major component of Lewy bodies and Lewy neurites in the brain of patients affected by Parkinson's disease (PD), Lewy body dementia (LBD), and other synucleinopathies. It is mainly involved in the regulation of synaptic vesicle trafficking but can also control mitochondrial/endoplasmic reticulum (ER) homeostasis, lysosome/phagosome function, and cytoskeleton organization. Recent evidence supports that the pathological forms of α-synuclein can also reduce the release of vasoactive and inflammatory mediators from endothelial cells (ECs) and modulates the expression of tight junction (TJ) proteins important for maintaining the blood-brain barrier (BBB). This hints that α-synuclein deposition can affect BBB integrity. Border associated macrophages (BAMs) are brain resident macrophages found in association with the vasculature (PVMs), meninges (MAMs), and choroid plexus (CPMs). Recent findings indicate that these cells play distinct roles in stroke and neurodegenerative disorders. Although many studies have addressed how α-synuclein may modulate microglia, its effect on BAMs has been scarcely investigated. This review aims at summarizing the main findings supporting how α-synuclein can affect ECs and/or BAMs function as well as their interplay and effect on other cells in the brain perivascular environment in physiological and pathological conditions. Gaps of knowledge and new perspectives on how this protein can contribute to neurodegeneration by inducing BBB homeostatic changes in different neurological conditions are highlighted.
Collapse
Affiliation(s)
- Tizibt Ashine Bogale
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gaia Faustini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesca Longhena
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Stefania Mitola
- Biotechnology Division, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Laboratory for Preventive and Personalized Medicine, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marina Pizzi
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Arianna Bellucci
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Laboratory for Preventive and Personalized Medicine, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
28
|
Chamera K, Trojan E, Kotarska K, Szuster-Głuszczak M, Bryniarska N, Tylek K, Basta-Kaim A. Role of Polyinosinic:Polycytidylic Acid-Induced Maternal Immune Activation and Subsequent Immune Challenge in the Behaviour and Microglial Cell Trajectory in Adult Offspring: A Study of the Neurodevelopmental Model of Schizophrenia. Int J Mol Sci 2021; 22:ijms22041558. [PMID: 33557113 PMCID: PMC7913889 DOI: 10.3390/ijms22041558] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/22/2021] [Accepted: 02/01/2021] [Indexed: 12/21/2022] Open
Abstract
Multiple lines of evidence support the pathogenic role of maternal immune activation (MIA) in the occurrence of the schizophrenia-like disturbances in offspring. While in the brain the homeostatic role of neuron-microglia protein systems is well documented, the participation of the CX3CL1-CX3CR1 and CD200-CD200R dyads in the adverse impact of MIA often goes under-recognized. Therefore, in the present study, we examined the effect of MIA induced by polyinosinic:polycytidylic acid (Poly I:C) on the CX3CL1-CX3CR1 and CD200-CD200R axes, microglial trajectory (MhcII, Cd40, iNos, Il-1β, Tnf-α, Il-6, Arg1, Igf-1, Tgf-β and Il-4), and schizophrenia-like behaviour in adult male offspring of Sprague-Dawley rats. Additionally, according to the “two-hit” hypothesis of schizophrenia, we evaluated the influence of acute challenge with Poly I:C in adult prenatally MIA-exposed animals on the above parameters. In the present study, MIA evoked by Poly I:C injection in the late period of gestation led to the appearance of schizophrenia-like disturbances in adult offspring. Our results revealed the deficits manifested as a diminished number of aggressive interactions, presence of depressive-like episodes, and increase of exploratory activity, as well as a dichotomy in the sensorimotor gating in the prepulse inhibition (PPI) test expressed as two behavioural phenotypes (MIAPPI-low and MIAPPI-high). Furthermore, in the offspring rats subjected to a prenatal challenge (i.e., MIA) we noticed the lack of modulation of behavioural changes after the additional acute immune stimulus (Poly I:C) in adulthood. The important finding reported in this article is that MIA affects the expression and levels of the neuron-microglia proteins in the frontal cortex and hippocampus of adult offspring. We found that the changes in the CX3CL1-CX3CR1 axis could affect microglial trajectory, including decreased hippocampal mRNA level of MhcII and elevated cortical expression of Igf-1 in the MIAPPI-high animals and/or could cause the up-regulation of an inflammatory response (Il-6, Tnf-α, iNos) after the “second hit” in both examined brain regions and, at least in part, might differentiate behavioural disturbances in adult offspring. Consequently, the future effort to identify the biological background of these interactions in the Poly I:C-induced MIA model in Sprague-Dawley rats is desirable to unequivocally clarify this issue.
Collapse
|
29
|
Timmerman R, Burm SM, Bajramovic JJ. Tissue-specific features of microglial innate immune responses. Neurochem Int 2020; 142:104924. [PMID: 33248205 DOI: 10.1016/j.neuint.2020.104924] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/21/2020] [Accepted: 11/22/2020] [Indexed: 02/07/2023]
Abstract
As tissue-resident macrophages of the brain, microglia are increasingly considered as cellular targets for therapeutical intervention. Innate immune responses in particular have been implicated in central nervous system (CNS) infections, neuro-oncology, neuroinflammatory and neurodegenerative diseases. We here review the impact of 'nature and nurture' on microglial innate immune responses and summarize documented tissue-specific adaptations. Overall, such adaptations are associated with regulatory processes rather than with overt differences in the expressed repertoire of activating receptors of different tissue-resident macrophages. Microglial responses are characterized by slower kinetics, by a more persistent nature and by a differential usage of downstream enzymes and accessory receptors. We further consider factors like aging, previous exposure to inflammatory stimuli, and differences in the microenvironment that can modulate innate immune responses. The long-life span of microglia in the metabolically active CNS renders them susceptible to the phenomenon of 'inflammaging', and major challenges lie in the unraveling of the factors that underlie age-related alterations in microglial behavior.
Collapse
Affiliation(s)
- R Timmerman
- Alternatives Unit, Biomedical Primate Research Centre, Rijswijk, the Netherlands
| | - S M Burm
- Genmab, Utrecht, the Netherlands
| | - J J Bajramovic
- Alternatives Unit, Biomedical Primate Research Centre, Rijswijk, the Netherlands.
| |
Collapse
|
30
|
Chamera K, Kotarska K, Szuster-Głuszczak M, Trojan E, Skórkowska A, Pomierny B, Krzyżanowska W, Bryniarska N, Basta-Kaim A. The prenatal challenge with lipopolysaccharide and polyinosinic:polycytidylic acid disrupts CX3CL1-CX3CR1 and CD200-CD200R signalling in the brains of male rat offspring: a link to schizophrenia-like behaviours. J Neuroinflammation 2020; 17:247. [PMID: 32829711 PMCID: PMC7444338 DOI: 10.1186/s12974-020-01923-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/10/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The bidirectional communication between neurons and microglia is fundamental for the homeostasis and biological function of the central nervous system. Maternal immune activation (MIA) is considered to be one of the factors affecting these interactions. Accordingly, MIA has been suggested to be involved in several neuropsychiatric diseases, including schizophrenia. The crucial regulatory systems for neuron-microglia crosstalk are the CX3CL1-CX3CR1 and CD200-CD200R axes. METHODS We aimed to clarify the impact of MIA on CX3CL1-CX3CR1 and CD200-CD200R signalling pathways in the brains of male Wistar rats in early and adult life by employing two neurodevelopmental models of schizophrenia based on the prenatal challenge with lipopolysaccharide (LPS) and polyinosinic:polycytidylic acid (Poly I:C). We also examined the effect of MIA on the expression of microglial markers and the profile of cytokines released in the brains of young offspring, as well as the behaviour of adult animals. Moreover, we visualized the localization of ligand-receptor systems in the hippocampal regions (CA1, CA3 and DG) and the frontal cortex of young rats exposed to MIA. The differences between groups were analysed using Student's t test. RESULTS We observed that MIA altered developmental trajectories in neuron-microglia communication in the brains of young offspring, as evidenced by the disruption of CX3CL1-CX3CR1 and/or CD200-CD200R axes. Our data demonstrated the presence of abnormalities after LPS-induced MIA in levels of Cd40, Il-1β, Tnf-α, Arg1, Tgf-β and Il-10, as well as IBA1, IL-1β and IL-4, while after Poly I:C-generated MIA in levels of Cd40, iNos, Il-6, Tgf-β, Il-10, and IBA1, IL-1β, TNF-α, IL-6, TGF-β and IL-4 early in the life of male animals. In adult male rats that experienced prenatal exposure to MIA, we observed behavioural changes resembling a schizophrenia-like phenotype. CONCLUSIONS Our study provides evidence that altered CX3CL1-CX3CR1 and/or CD200-CD200R pathways, emerging after prenatal immune challenge with LPS and Poly I:C, might be involved in the aetiology of schizophrenia.
Collapse
Affiliation(s)
- Katarzyna Chamera
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Katarzyna Kotarska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Magdalena Szuster-Głuszczak
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Ewa Trojan
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Alicja Skórkowska
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Collegium Medicum, 9 Medyczna St, 30-688, Kraków, Poland
| | - Bartosz Pomierny
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Collegium Medicum, 9 Medyczna St, 30-688, Kraków, Poland
| | - Weronika Krzyżanowska
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Collegium Medicum, 9 Medyczna St, 30-688, Kraków, Poland
| | - Natalia Bryniarska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland.
| |
Collapse
|
31
|
Maternal Immune Activation Sensitizes Male Offspring Rats to Lipopolysaccharide-Induced Microglial Deficits Involving the Dysfunction of CD200-CD200R and CX3CL1-CX3CR1 Systems. Cells 2020; 9:cells9071676. [PMID: 32664639 PMCID: PMC7407118 DOI: 10.3390/cells9071676] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/09/2020] [Accepted: 07/09/2020] [Indexed: 02/06/2023] Open
Abstract
Early life challenges resulting from maternal immune activation (MIA) may exert persistent effects on the offspring, including the development of psychiatric disorders, such as schizophrenia. Recent evidence has suggested that the adverse effects of MIA may be mediated by neuron-microglia crosstalk, particularly CX3CL1-CX3CR1 and CD200-CD200R dyads. Therefore, the present study assessed the behavioural parameters resembling schizophrenia-like symptoms in the adult male offspring of Sprague-Dawley rats that were exposed to MIA and to an additional acute lipopolysaccharide (LPS) challenge in adulthood, according to the "two-hit" hypothesis of schizophrenia. Simultaneously, we aimed to clarify the role of the CX3CL1-CX3CR1 and CD200-CD200R axes and microglial reactivity in the brains of adult offspring subjected to MIA and the "second hit" wit LPS. In the present study, MIA generated a range of behavioural changes in the adult male offspring, including increased exploratory activity and anxiety-like behaviours. The most intriguing finding was observed in the prepulse inhibition (PPI) test, where the deficit in the sensorimotor gating was age-dependent and present only in part of the rats. We were able to distinguish the occurrence of two groups: responsive and non-responsive (without the deficit). Concurrently, based on the results of the biochemical studies, MIA disrupted mainly the CD200-CD200R system, while the changes of the CX3CL1-CX3CR1 axis were less evident in the frontal cortex of adult non-responsive offspring. MIA markedly affected the immune regulators of the CD200-CD200R pathway as we observed an increase in cortical IL-6 release in the responsive group and IL-4 in the non-responsive offspring. Importantly, the "second hit" generated disturbances at the behavioural and biochemical levels mostly in the non-responsive adult animals. Those offspring were characterized both by disturbed PPI and "priming" microglia. Altogether, the exposure to MIA altered the immunomodulatory mechanisms, including the CD200-CD200R axis, in the brain and sensitized animals to subsequent immunological challenges, leading to the manifestation of schizophrenia-like alterations.
Collapse
|
32
|
Sun H, He X, Tao X, Hou T, Chen M, He M, Liao H. The CD200/CD200R signaling pathway contributes to spontaneous functional recovery by enhancing synaptic plasticity after stroke. J Neuroinflammation 2020; 17:171. [PMID: 32473633 PMCID: PMC7260848 DOI: 10.1186/s12974-020-01845-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 05/19/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Spontaneous functional recovery occurs during the acute phase after stroke onset, but this intrinsic recovery remains limited. Therefore, exploring the mechanism underlying spontaneous recovery and identifying potential strategies to promote functional rehabilitation after stroke are very important. The CD200/CD200R signaling pathway plays an important role in neurological recovery by modulating synaptic plasticity during multiple brain disorders. However, the effect and mechanism of action of the CD200/CD200R pathway in spontaneous functional recovery after stroke are unclear. METHODS In this study, we used a transient middle cerebral artery occlusion (MCAO) model in rats to investigate the function of CD200/CD200R signaling in spontaneous functional recovery after stroke. We performed a battery of behavioral tests (Longa test, adhesive removal test, limb-use asymmetry test, and the modified grip-traction test) to evaluate sensorimotor function after intracerebroventricular (i.c.v.) injection with CD200 fusion protein (CD200Fc) or CD200R blocking antibody (CD200R Ab) post-stroke. Density and morphology of dendritic spines were analyzed by Golgi staining. Microglia activation was evaluated by immunofluorescence staining. Western blot was used to detect the levels of protein and the levels of mRNA were measured by qPCR. RESULTS Our study demonstrated that sensorimotor function, synaptic proteins, and structures were gradually recovered and CD200R was transiently upregulated in ipsilateral cortex after stroke. Synapse-related proteins and dendritic spines were preserved, accompanied by sensorimotor functional recovery, after stereotaxic CD200Fc injection post-stroke. In addition, CD200Fc restrained microglia activation and pro-inflammatory factor release (such as Il-1, Tnf-α, and Il-6) after MCAO. On the contrary, CD200R Ab aggravated sensory function recovery in adhesive removal test and further promoted microglia activation and pro-inflammatory factor release (such as Il-1) after MCAO. The immune-modulatory effect of CD200/CD200R signaling might be exerted partly by its inhibition of the MAPK pathway. CONCLUSIONS This study provides evidence that the CD200/CD200R signaling pathway contributes to spontaneous functional recovery by enhancing synaptic plasticity via inhibition of microglia activation and inflammatory factor release.
Collapse
Affiliation(s)
- Hao Sun
- Jiangsu Key laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing, 210009, China
| | - Xinran He
- Jiangsu Key laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing, 210009, China
| | - Xia Tao
- Jiangsu Key laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing, 210009, China
| | - Tingting Hou
- Jiangsu Key laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing, 210009, China
| | - Mingming Chen
- Jiangsu Key laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing, 210009, China
| | - Meijun He
- Jiangsu Key laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing, 210009, China
| | - Hong Liao
- Jiangsu Key laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing, 210009, China.
| |
Collapse
|
33
|
Lin S, He L, Shen R, Fang F, Pan H, Zhu X, Wang M, Zhou Z, Liu Z, Wang X, Fang S, Sun X, Wang Y, Chen S, Ding J. Identification of the CD200R1 promoter and the association of its polymorphisms with the risk of Parkinson's disease. Eur J Neurol 2020; 27:1224-1230. [PMID: 32190938 DOI: 10.1111/ene.14224] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/04/2020] [Accepted: 03/05/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND AND PURPOSE Neuroinflammation is known to be involved in the pathogenesis of Parkinson's disease (PD). Abnormal activation of microglia plays a key role in this pathological process. CD200R1 is a membrane glycoprotein that is expressed primarily on myeloid cells including microglia and is involved in the maintenance of microglia in a stationary state. Our previous study reported that the regulation of CD200R1 expression is altered in PD patients. Such alteration will lead to neuroinflammation and is related to the pathogenesis of PD. The possible role of promoter polymorphisms for abnormal CD200R1 expression in PD was examined in this study. METHOD The UCSC database and dual-luciferase assays were used to confirm the promoter region of CD200R1. The promoter of CD200R1 was sequenced in 457 PD patients and 520 matched healthy controls from the Chinese Han population. Dual-luciferase assays were conducted to examine the promoter activity of CD200R1. RESULTS It was confirmed that the promoter of CD200R1 is located in the region 876-146 bp upstream of the coding DNA sequence. The frequencies of rs144721913 (P = 0.001) and rs72952157 (P = 0.022) in the promoter were significantly different between the PD group and control group. rs144721913 increases the risk of PD by approximately 14-fold and rs72952157 by 2.6-fold. The dual-luciferase assay indicated that the rs144721913 T allele and the rs72952157 G allele reduced the transcriptional activity of the CD200R1 promoter. CONCLUSIONS For the first time the promoter region of CD200R1 has been defined and two potential risk polymorphisms (rs144721913 and rs72952157) in the region for PD in Chinese Han populations have been reported.
Collapse
Affiliation(s)
- S Lin
- Institute of Neurology and Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - L He
- Institute of Neurology and Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - R Shen
- Institute of Neurology and Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - F Fang
- Department of Aging, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - H Pan
- Institute of Neurology and Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - X Zhu
- Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - M Wang
- Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Z Zhou
- Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Z Liu
- Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - X Wang
- Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, Fujian Provincial Maternity and Children Health Hospital, Affiliated Hospital of Fujian Medical University, FuJian, China
| | - S Fang
- Department of Neuroscience, University of California San Diego, La Jolla, California, USA
| | - X Sun
- Institute of Neurology and Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Y Wang
- Institute of Neurology and Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - S Chen
- Institute of Neurology and Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - J Ding
- Institute of Neurology and Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Wang L, Liu Y, Yan S, Du T, Fu X, Gong X, Zhou X, Zhang T, Wang X. Disease Progression-Dependent Expression of CD200R1 and CX3CR1 in Mouse Models of Parkinson's Disease. Aging Dis 2020; 11:254-268. [PMID: 32257540 PMCID: PMC7069458 DOI: 10.14336/ad.2019.0615] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/15/2019] [Indexed: 12/16/2022] Open
Abstract
Microglial activation is an important contributor to the pathogenesis of Parkinson’s disease (PD). Microglia are tightly and efficiently regulated by immune checkpoints, including CD200-CD200R1 and CX3CL1-CX3CR1. Understanding the involvement of these checkpoints in disease progression provides important insights into how microglial activation contributes to PD pathology. However, so far, studies have produced seemingly conflicting results. In this study, we demonstrate that CD200R1 expression is down-regulated at both early and late stage of PD model, and CX3CR1 expression is down-regulated in early stage and recovered in late stage. In primary cultured microglia, CD200R1 and CX3CR1 expressions are both directly regulated by LPS or α-synuclein, and CD200R1 expression is more sensitively regulated than CX3CR1. In addition, CD200 knockout causes an increase in proinflammatory cytokine production and microglial activation in the midbrain. Remarkably, DA neurons in the substantial nigra are degenerated in CD200-/- mice. Finally, activation of the CD200R with CD200Fc alleviates the neuroinflammation in microglia. Together, these results suggest that immune checkpoints play distinct functional roles in different stage of PD pathology, and the CD200-CD200R1 axis plays a significant role in nigrostriatal neuron viability and function.
Collapse
Affiliation(s)
- Le Wang
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Yang Liu
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Shuxin Yan
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Tianshu Du
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xia Fu
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xiaoli Gong
- 2Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| | - Xinyu Zhou
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Ting Zhang
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xiaomin Wang
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China.,2Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| |
Collapse
|
35
|
Wang L, Gong X, Liu Y, Du T, Zhang Z, Zhang T, Wang X. CD200 maintains the region-specific phenotype of microglia in the midbrain and its role in Parkinson's disease. Glia 2020; 68:1874-1890. [PMID: 32112601 DOI: 10.1002/glia.23811] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/23/2020] [Accepted: 02/18/2020] [Indexed: 12/21/2022]
Abstract
Microglia are a specialized population of tissue macrophages in the mammalian brain. Microglial phenotype is tightly regulated by local environmental factors, although little is known about these factors and their region-preferred roles in regulating local neuroinflammatory responses. We hypothesized that microglia in different brain regions respond differently to neuroinflammatory stimulation and that CD200, an anti-inflammatory protein mainly originated from neurons, acts as a local cue inhibiting microglia activation in the midbrain. We utilized a CD200-deficient mouse line to analyze the phenotypic role of CD200 in the regulation of normal neuron-microglia homeostasis in the midbrain and in the dopaminergic degeneration in an α-synuclein overexpression model of PD. We found that systemic administration of an endotoxin lipopolysaccharide induced a region-preferred change in CD200 expression in the midbrain. Similarly, CD200-/- mice showed a regional preference in an enhancement of microglia activation and baseline inflammatory levels in the midbrain and dopamine neuron loss in the substantia nigra (SN). In a mouse model of Parkinson's disease (PD) induced by rAAV-hSYN injection into the SN, CD200-/- mice showed more dopamine neuron loss in the SN than wild type mice. Activation of CD200 receptors with a CD200 fusion protein alleviated the neuroinflammation and neuronal death in the SN of PD mice. These findings demonstrate that CD200 is essential for the midbrain homeostasis and acts as a critical local regulator in controlling microglial properties related to the PD pathogenesis.
Collapse
Affiliation(s)
- Le Wang
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xiaoli Gong
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| | - Yang Liu
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Tianshu Du
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Zhen Zhang
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Ting Zhang
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xiaomin Wang
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| |
Collapse
|
36
|
Yin P, Wang S, Wei Y, Wang X, Zhang J, Yin X, Feng J, Zhu M. Maresin1 Decreased Microglial Chemotaxis and Ameliorated Inflammation Induced by Amyloid-β42 in Neuron-Microglia Co-Culture Models. J Alzheimers Dis 2020; 73:503-515. [PMID: 31796671 DOI: 10.3233/jad-190682] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Ping Yin
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
- Department of Neurology, Heilongjiang Provincial Hospital, Harbin, China
| | - Shuang Wang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yafen Wei
- Department of Neurology, Heilongjiang Provincial Hospital, Harbin, China
| | - Xu Wang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Jingdian Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Xiang Yin
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Jiachun Feng
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Mingqin Zhu
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
37
|
Nielsen JJJ, Lillethorup TP, Glud AN, Sørensen JCH, Orlowski D. The application of iPSCs in Parkinson’s disease. Acta Neurobiol Exp (Wars) 2020. [DOI: 10.21307/ane-2020-024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
38
|
Chamera K, Trojan E, Szuster-Głuszczak M, Basta-Kaim A. The Potential Role of Dysfunctions in Neuron-Microglia Communication in the Pathogenesis of Brain Disorders. Curr Neuropharmacol 2020; 18:408-430. [PMID: 31729301 PMCID: PMC7457436 DOI: 10.2174/1570159x17666191113101629] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/15/2019] [Accepted: 11/10/2019] [Indexed: 12/18/2022] Open
Abstract
The bidirectional communication between neurons and microglia is fundamental for the proper functioning of the central nervous system (CNS). Chemokines and clusters of differentiation (CD) along with their receptors represent ligand-receptor signalling that is uniquely important for neuron - microglia communication. Among these molecules, CX3CL1 (fractalkine) and CD200 (OX-2 membrane glycoprotein) come to the fore because of their cell-type-specific localization. They are principally expressed by neurons when their receptors, CX3CR1 and CD200R, respectively, are predominantly present on the microglia, resulting in the specific axis which maintains the CNS homeostasis. Disruptions to this balance are suggested as contributors or even the basis for many neurological diseases. In this review, we discuss the roles of CX3CL1, CD200 and their receptors in both physiological and pathological processes within the CNS. We want to underline the critical involvement of these molecules in controlling neuron - microglia communication, noting that dysfunctions in their interactions constitute a key factor in severe neurological diseases, such as schizophrenia, depression and neurodegeneration-based conditions.
Collapse
Affiliation(s)
- Katarzyna Chamera
- Department of Experimental Neuroendocrinology, Laboratory of Immunoendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St. 31-343Kraków, Poland
| | - Ewa Trojan
- Department of Experimental Neuroendocrinology, Laboratory of Immunoendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St. 31-343Kraków, Poland
| | - Magdalena Szuster-Głuszczak
- Department of Experimental Neuroendocrinology, Laboratory of Immunoendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St. 31-343Kraków, Poland
| | - Agnieszka Basta-Kaim
- Department of Experimental Neuroendocrinology, Laboratory of Immunoendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St. 31-343Kraków, Poland
| |
Collapse
|
39
|
Ahmad W, Ali A, Ali A, Khan S, Khan S, Husain I. Upcoming diagnostic biomarkers with promising prospects in neurological disorders. Clin Exp Pharmacol Physiol 2019; 47:347-356. [PMID: 31746003 DOI: 10.1111/1440-1681.13216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 09/09/2019] [Accepted: 11/16/2019] [Indexed: 12/12/2022]
Abstract
An exponential increase in the prevalence of neurological disorders requires substantial steps to be taken for their prevention and treatment. Neurodiagnostic biomarkers are gaining momentum presently in order to enhance the diagnostic accuracy of neurodegenerative disorders, to precisely assess their advancement and to monitor the efficiency of therapeutic interventions. Therefore, the primary focus of the present review is the recent development in this field of neurodiagnostic biomarkers, and the current state of biomarker exploration in the context of various neurodegenerative diseases. This review encompasses an updated and detailed account of specific (β-Amyloid, Tau and Phospho-tau 181, Tar-DNA binding protein-43, Progranulin, a-synuclein, Clusterin, etc) and non-specific (genetic, synaptic, inflammatory and coagulation) neurodiagnostic biomarkers and the recent advances in this growing field. This comprehensive review also suggests the utilization of neurodiagnostic markers in network approaches and personalized medication that will eventually improvise the existing diagnostic and therapeutic complexities of neurodiagnostic biomarkers.
Collapse
Affiliation(s)
- Wasim Ahmad
- Department of Pharmacy, Mohammad Al-Mana College for Medical Sciences, Dammam, Saudi Arabia
| | - Abuzer Ali
- College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Amena Ali
- College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Sana Khan
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, India
| | - Saba Khan
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, India
| | | |
Collapse
|
40
|
Ngwa C, Liu F. CD200-CD200R signaling and diseases: a potential therapeutic target? INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2019; 11:297-309. [PMID: 31993106 PMCID: PMC6971504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 12/13/2019] [Indexed: 06/10/2023]
Abstract
CD200 and its receptor, CD200R, constitutes an endogenous inhibitory signaling, and is being increasingly recognized in studies of various central nervous system (CNS) disorders. Emerging data have demonstrated that neuronal CD200 binds to CD200R to modulate immune responses to pathogenic stimuli. However, on which component of the immune response that CD200-CD200R signaling acts is not well understood. In this review, we focused on cellular expression of the signaling, the effects on immune cell activation, and the function in pathological procedures of neurodegenerative diseases, in both clinical and experimental disease models. Essential functions of CD200-CD200R interaction and the treatment relevance have been elaborated. Immune responses to diseases under the control of CD200-CD200R axis were also discussed in the review.
Collapse
Affiliation(s)
- Conelius Ngwa
- Department of Neurology, The University of Texas Health Science Center at Houston McGovern Medical School Houston, TX 77030, USA
| | - Fudong Liu
- Department of Neurology, The University of Texas Health Science Center at Houston McGovern Medical School Houston, TX 77030, USA
| |
Collapse
|
41
|
Liu CY, Wang X, Liu C, Zhang HL. Pharmacological Targeting of Microglial Activation: New Therapeutic Approach. Front Cell Neurosci 2019; 13:514. [PMID: 31803024 PMCID: PMC6877505 DOI: 10.3389/fncel.2019.00514] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 10/31/2019] [Indexed: 12/13/2022] Open
Abstract
Mounting evidence suggests that neuroinflammation is not just a consequence but a vital contributor to the development and progression of Parkinson’s disease (PD). Microglia in particular, may contribute to the induction and modulation of inflammation in PD. Upon stimulation, microglia convert into activated phenotypes, which exist along a dynamic continuum and bear different immune properties depending on the disease stage and severity. Activated microglia release various factors involved in neuroinflammation, such as cytokines, chemokines, growth factors, reactive oxygen species (ROS), reactive nitrogen species (RNS), and prostaglandins (PGs). Further, activated microglia interact with other cell types (e.g., neurons, astrocytes and mast cells) and are closely associated with α-synuclein (α-syn) pathophysiology and iron homeostasis disturbance. Taken together, microglial activation and microglia-mediated inflammatory responses play essential roles in the pathogenesis of PD and elucidation of the complexity and imbalance of microglial activation may shed light on novel therapeutic approaches for PD.
Collapse
Affiliation(s)
- Cai-Yun Liu
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Xu Wang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Chang Liu
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Hong-Liang Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Department of Life Sciences, National Natural Science Foundation of China, Beijing, China
| |
Collapse
|
42
|
Sun H, Li A, Hou T, Tao X, Chen M, Wu C, Chen S, Zhu L, Liao H. Neurogenesis promoted by the CD200/CD200R signaling pathway following treadmill exercise enhances post-stroke functional recovery in rats. Brain Behav Immun 2019; 82:354-371. [PMID: 31513876 DOI: 10.1016/j.bbi.2019.09.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/22/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022] Open
Abstract
Stroke is a leading cause of long-term disability worldwide; survivors often show sensorimotor and cognitive deficits. Therapeutic exercise is the most common treatment strategy for rehabilitating patients with stroke via augmentation of neurogenesis, angiogenesis, neurotrophic factors expression, and synaptogenesis. Neurogenesis plays important roles in sensorimotor and cognitive functional recovery, and can be promoted by exercise; however, the mechanism underlying this phenomenon remains unclear. In this study, we explored the effects of treadmill exercise on sensorimotor and cognitive functional recovery, as well as the potential molecular mechanisms underlying the promotion of neurogenesis in a rat model of transient middle cerebral artery occlusion (tMCAO). We found that treadmill exercise facilitated sensorimotor and cognitive functional recovery after tMCAO, and that neural stem/progenitor cell proliferation, differentiation, and migration were enhanced in the ipsilateral subventricular and subgranular zones after tMCAO. Meanwhile, the newborn neurons induced by treadmill exercise after tMCAO had the similar function with pre-existing neurons. Treadmill exercise significantly increased CD200 and CD200 receptor (CD200R) levels in the ipsilateral hippocampus and cortex. Further study revealed that treadmill exercise-induced neurogenesis and functional recovery were clearly inhibited, while Il-β and Tnf-α expression were upregulated, following lentivirus (LV)-induced suppression of post-stroke CD200R expression. Consistent with the effect of treadmill exercise, CD200Fc (a CD200R agonist) markedly promoted neurogenesis and functional recovery after stroke. In addition, CD200Fc could further enhance the functional recovery induced by treadmill exercise after stroke. Our results demonstrate the beneficial role of treadmill exercise in promoting neurogenesis and functional recovery via activating the CD200/CD200R signaling pathway and improving the inflammatory environment after stroke. Thus, the CD200/CD200R signaling pathway is a potential therapeutic target for functional recovery after stroke.
Collapse
Affiliation(s)
- Hao Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China
| | - Ao Li
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China
| | - Tingting Hou
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China
| | - Xia Tao
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China
| | - Mingming Chen
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China
| | - Chaoran Wu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China
| | - Shujian Chen
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China
| | - Lingling Zhu
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.
| | - Hong Liao
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China; State Key Laboratory of National Medicines, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China.
| |
Collapse
|
43
|
Zhao X, Li J, Sun H. CD200-CD200R Interaction: An Important Regulator After Stroke. Front Neurosci 2019; 13:840. [PMID: 31440137 PMCID: PMC6693438 DOI: 10.3389/fnins.2019.00840] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/26/2019] [Indexed: 12/16/2022] Open
Abstract
The high mortality and morbidity rate of stroke is a chronic problem that plagues human society. The activation of microglia is one of the principal reasons why neuroinflammation induces cerebral dysfunction. Because of their vital functions in the regulation of neuroinflammation, microglia constitute an important target for stroke. Given that there is an innate self-preservation mechanism between neurons and microglia, the transmembrane glycoproteins on the surface of their membranes, namely CD200 and CD200R, have become a popular topic of research. Numerous studies have demonstrated that CD200-CD200R interaction, microglial activation, and poststroke neuroinflammatory damage are inextricably linked. In this review, we describe the above relationship from a new perspective. We specifically focus on neuroinflammation after stroke. The role of crosstalk of CD200-CD200R inhibitory immune ligand receptors in immune regulation will also be illustrated. Thus, we will see how poststroke injury can be influenced by the CD200-CD200R crosstalk. Finally, we will discuss the possibility of clinical application of the result of CD200-CD200R interaction to manage neuroinflammatory injury after stroke.
Collapse
Affiliation(s)
- Xu Zhao
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,The Second Clinical Medical College, Southern Medical University, Guangzhou, China.,Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brian Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| | - Jing Li
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,The Second Clinical Medical College, Southern Medical University, Guangzhou, China.,Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brian Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| | - Haitao Sun
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,The Second Clinical Medical College, Southern Medical University, Guangzhou, China.,Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brian Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| |
Collapse
|
44
|
Mee-Inta O, Zhao ZW, Kuo YM. Physical Exercise Inhibits Inflammation and Microglial Activation. Cells 2019; 8:cells8070691. [PMID: 31324021 PMCID: PMC6678635 DOI: 10.3390/cells8070691] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 12/20/2022] Open
Abstract
Accumulating evidence indicates that exercise can enhance brain function and attenuate neurodegeneration. Besides improving neuroplasticity by altering the synaptic structure and function in various brain regions, exercise also modulates multiple systems that are known to regulate neuroinflammation and glial activation. Activated microglia and several pro-inflammatory cytokines play active roles in the pathogenesis of neurodegenerative diseases, such as Alzheimer’s disease and Parkinson’s disease. The purpose of this review is to highlight the impacts of exercise on microglial activation. Possible mechanisms involved in exercise-modulated microglial activation are also discussed. Undoubtedly, more studies are needed in order to disclose the detailed mechanisms, but this approach offers therapeutic potential for improving the brain health of millions of aging people where pharmacological intervention has failed.
Collapse
Affiliation(s)
- Onanong Mee-Inta
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 70101, Taiwan
| | - Zi-Wei Zhao
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yu-Min Kuo
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 70101, Taiwan.
- Department of Cell Biology and Anatomy, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
45
|
Feng D, Huang A, Yan W, Chen D. CD200 dysfunction in neuron contributes to synaptic deficits and cognitive impairment. Biochem Biophys Res Commun 2019; 516:1053-1059. [PMID: 31277944 DOI: 10.1016/j.bbrc.2019.06.134] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 06/23/2019] [Indexed: 10/26/2022]
Abstract
Neuronal CD200 plays critical role in neuron-microglia communication and negatively related to pathological development of Alzheimer's disease, but its contribution retroacts to synaptic function is still not clear. Here, we directly injected adeno-associated virus into hippocampus of APP/PS1 mice to induce neuron-specific CD200 overexpression; subsequently, we detected cognition function, synaptic function, and the microglial responses during AD pathological development. Notably, specific upregulation of neuronal CD200 promoted cognitive function by inhibiting microglial activation and secretion, improving synaptic function, and preventing synaptic loss. To further investigate the role of neuronal CD200 on synaptic function, we depressed CD200 expression in neuron by hippocampal AAV injection and also evaluated microglial activation and cognitive and synaptic function. Interestingly, data revealed that neuronal CD200 deficiency significantly impairs synaptic and cognitive function. Taken these results together, we concluded that neuronal CD200 mediated microglial response and synaptic function during AD pathological development, and provided evidence that neuronal CD200 restoration ameliorates synaptic and cognitive deficits.
Collapse
Affiliation(s)
- Depeng Feng
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China, 252000
| | - Anqi Huang
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China, 252000
| | - Wei Yan
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China, 252000.
| | - Dezhe Chen
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China, 252000
| |
Collapse
|
46
|
Microglia-neuron crosstalk: Signaling mechanism and control of synaptic transmission. Semin Cell Dev Biol 2019; 94:138-151. [PMID: 31112798 DOI: 10.1016/j.semcdb.2019.05.017] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/17/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022]
Abstract
The continuous crosstalk between microglia and neurons is required for microglia housekeeping functions and contributes to brain homeostasis. Through these exchanges, microglia take part in crucial brain functions, including development and plasticity. The alteration of neuron-microglia communication contributes to brain disease states with consequences, ranging from synaptic function to neuronal survival. This review focuses on the signaling pathways responsible for neuron-microglia crosstalk, highlighting their physiological roles and their alteration or specific involvement in disease. In particular, we discuss studies, establishing how these signaling allow microglial cells to control relevant physiological functions during brain development, including synaptic formation and circuit refinement. In addition, we highlight how microglia and neurons interact functionally to regulate highly dynamical synaptic functions. Microglia are able to release several signaling molecules involved in the regulation of synaptic activity and plasticity. On the other side, molecules of neuronal origin control microglial processes motility in an activity-dependent manner. Indeed, the continuous crosstalk between microglia and neurons is required for the sensing and housekeeping functions of microglia and contributes to the maintenance of brain homeostasis and, particularly, to the sculpting of neuronal connections during development. These interactions lay on the delicate edge between physiological processes and homeostasis alteration in pathology and are themselves altered during neuroinflammation. The full description of these processes could be fundamental for understanding brain functioning in health and disease.
Collapse
|
47
|
Jones MK, Nair A, Gupta M. Mast Cells in Neurodegenerative Disease. Front Cell Neurosci 2019; 13:171. [PMID: 31133804 PMCID: PMC6524694 DOI: 10.3389/fncel.2019.00171] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 04/11/2019] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases affect millions of people worldwide, yet there are currently no effective treatments. Because risk of neurodegenerative disease substantially increases with age, greater life expectancy with a concomitant aging population means more individuals will be affected in the coming decades. Thus, there is an urgent need for understanding the mechanisms driving neurodegenerative diseases in order to develop improved treatment strategies. Inflammation in the nervous system, termed “neuroinflammation,” has become increasingly recognized as being associated with neurodegenerative diseases. Early attention focused primarily on morphological changes in astrocytes and microglia; however, brain and CNS resident mast cells are now receiving attention as a result of being “first responders” to injury. Mast cells also exert profound effects on their microenvironment and neighboring cells including behavior and/or activation of astrocytes, microglia, and neurons, which, in turn, are implicated in neuroinflammation, neurogenesis and neurodegeneration. Mast cells also affect disruption/permeability of the blood brain barrier enabling toxin and immune cell entry exacerbating an inflammatory microenvironment. Here, we discuss the roles of mast cells in neuroinflammation and neurodegeneration with a focus on development and progression of four prominent neurodegenerative diseases: Alzheimer’s Disease, Parkinson’s Disease, Amyotrophic Lateral Sclerosis, and Huntington’s Disease.
Collapse
Affiliation(s)
- Michael K Jones
- Department of Medicine, Vascular Biology Center, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - Archana Nair
- Department of Ophthalmology, New York University, New York, NY, United States
| | - Mihir Gupta
- Department of Neurosurgery, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
48
|
Garretti F, Agalliu D, Lindestam Arlehamn CS, Sette A, Sulzer D. Autoimmunity in Parkinson's Disease: The Role of α-Synuclein-Specific T Cells. Front Immunol 2019; 10:303. [PMID: 30858851 PMCID: PMC6397885 DOI: 10.3389/fimmu.2019.00303] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/06/2019] [Indexed: 12/23/2022] Open
Abstract
Evidence from a variety of studies implicates a role for the adaptive immune system in Parkinson's disease (PD). Similar to multiple sclerosis (MS) patients who display a high number of T cells in the brain attacking oligodendrocytes, PD patients show higher numbers of T cells in the ventral midbrain than healthy, age-matched controls. Mouse models of the disease also show the presence of T cells in the brain. The role of these infiltrating T cells in the propagation of disease is controversial; however, recent studies indicate that they may be autoreactive in nature, recognizing disease-altered self-proteins as foreign antigens. T cells of PD patients can generate an autoimmune response to α-synuclein, a protein that is aggregated in PD. α-Synuclein and other proteins are post-translationally modified in an environment in which protein processing is altered, possibly leading to the generation of neo-epitopes, or self-peptides that have not been identified by the host immune system as non-foreign. Infiltrating T cells may also be responding to such modified proteins. Genome-wide association studies (GWAS) have shown associations of PD with haplotypes of major histocompatibility complex (MHC) class II genes, and a polymorphism in a non-coding region that may increase MHC class II in PD patients. We speculate that the inflammation observed in PD may play both pathogenic and protective roles. Future studies on the adaptive immune system in neurodegenerative disorders may elucidate steps in disease pathogenesis and assist with the development of both biomarkers and treatments.
Collapse
Affiliation(s)
- Francesca Garretti
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States
| | - Dritan Agalliu
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States.,Department of Neurology, Columbia University Irving Medical Center, New York, NY, United States
| | | | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States.,Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - David Sulzer
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, United States.,Department of Pharmacology, Columbia University Irving Medical Center, New York, NY, United States.,Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
49
|
Feng Z, Ye L, Klebe D, Ding Y, Guo ZN, Flores JJ, Yin C, Tang J, Zhang JH. Anti-inflammation conferred by stimulation of CD200R1 via Dok1 pathway in rat microglia after germinal matrix hemorrhage. J Cereb Blood Flow Metab 2019; 39:97-107. [PMID: 28792282 PMCID: PMC6311673 DOI: 10.1177/0271678x17725211] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
CD200 has been reported to be neuroprotective in neurodegenerative diseases. However, the potential protective effects of CD200 in germinal matrix hemorrhage (GMH) have not been investigated. We examined the anti-inflammatory mechanisms of CD200 after GMH. A total of 167 seven-day-old rat pups were used. The time-dependent effect of GMH on the levels of CD200 and CD200 Receptor 1 (CD200R1) was evaluated by western blot. CD200R1 was localized by immunohistochemistry. The short-term (24 h) and long-term (28 days) outcomes were evaluated after CD200 fusion protein (CD200Fc) treatment by neurobehavioral assessment. CD200 small interfering RNA (siRNA) and downstream of tyrosine kinase 1 (Dok1) siRNA were injected intracerebroventricularly. Western blot was employed to study the mechanisms of CD200 and CD200R1. GMH induced significant developmental delay and caused impairment in both cognitive and motor functions in rat pups. CD200Fc ameliorated GMH-induced damage. CD200Fc increased expression of Dok1 and decreased IL-1beta and TNF-alpha levels. CD200R1 siRNA and Dok1 siRNA abolished the beneficial effects of CD200Fc, as demonstrated by enhanced expression levels of IL-1beta and TNF-alpha. CD200Fc inhibited GMH-induced inflammation and this effect may be mediated by CD200R1/Dok1 pathway. Thus, CD200Fc may serve as a potential treatment to ameliorate brain injury for GMH patients.
Collapse
Affiliation(s)
- Zhanhui Feng
- 1 Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, China.,2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Lan Ye
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.,3 The Medical Function Laboratory of Experimental Teaching Center of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Damon Klebe
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Yan Ding
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Zhen-Ni Guo
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Jerry J Flores
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Cheng Yin
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Jiping Tang
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.,4 Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, USA.,5 Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
50
|
Co-signaling Molecules in Neurological Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:233-265. [PMID: 31758537 DOI: 10.1007/978-981-32-9717-3_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|